Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397093

RESUMEN

The lung can experience different oxygen concentrations, low as in hypoxia, high as under supplemental oxygen therapy, or oscillating during intermittent hypoxia as in obstructive sleep apnea or intermittent hypoxia/hyperoxia due to cyclic atelectasis in the ventilated patient. This study aimed to characterize the oxygen-condition-specific protein composition of extracellular vesicles (EVs) released from human pulmonary microvascular endothelial cells in vitro to decipher their potential role in biotrauma using quantitative proteomics with bioinformatic evaluation, transmission electron microscopy, flow cytometry, and non-activated thromboelastometry (NATEM). The release of vesicles enriched in markers CD9/CD63/CD81 was enhanced under intermittent hypoxia, strong hyperoxia and intermittent hypoxia/hyperoxia. Particles with exposed phosphatidylserine were increased under intermittent hypoxia. A small portion of vesicles were tissue factor-positive, which was enhanced under intermittent hypoxia and intermittent hypoxia/hyperoxia. EVs from treatment with intermittent hypoxia induced a significant reduction of Clotting Time in NATEM analysis compared to EVs isolated after normoxic exposure, while after intermittent hypoxia/hyperoxia, tissue factor in EVs seems to be inactive. Gene set enrichment analysis of differentially expressed genes revealed that EVs from individual oxygen conditions potentially induce different biological processes such as an inflammatory response under strong hyperoxia and intermittent hypoxia/hyperoxia and enhancement of tumor invasiveness under intermittent hypoxia.


Asunto(s)
Vesículas Extracelulares , Hiperoxia , Humanos , Oxígeno/farmacología , Oxígeno/metabolismo , Hiperoxia/metabolismo , Proteoma/metabolismo , Células Endoteliales/patología , Tromboplastina/metabolismo , Pulmón/patología , Hipoxia/metabolismo , Vesículas Extracelulares/metabolismo , Endotelio/patología
2.
Crit Care Med ; 50(9): e696-e706, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35191411

RESUMEN

OBJECTIVES: Ventilator-induced lung injury (VILI) is a major contributor to morbidity and mortality in critically ill patients. Mechanical damage to the lungs is potentially aggravated by the activation of the renin-angiotensin system (RAS). This article describes RAS activation profiles in VILI and discusses the effects of angiotensin (Ang) 1-7 supplementation or angiotensin-converting enzyme (ACE) inhibition with captopril as protective strategies. DESIGN: Animal study. SETTING: University research laboratory. SUBJECTS: C57BL/6 mice. INTERVENTIONS: Anesthetized mice ( n = 12-18 per group) were mechanically ventilated with low tidal volume (LV T , 6 mL/kg), high tidal volume (HV T , 15 mL/kg), or very high tidal volume (VHV T , 30 mL/kg) for 4 hours, or killed after 3 minutes (sham). Additional VHV T groups received infusions of 60 µg/kg/hr Ang 1-7 or a single dose of 100 mg/kg captopril. MEASUREMENTS AND MAIN RESULTS: VILI was characterized by increased bronchoalveolar lavage fluid levels of interleukin (IL)-6, keratinocyte-derived cytokine, and macrophage inflammatory protein-2 (MIP2). The Ang metabolites in plasma measured with liquid chromatography tandem mass spectrometry showed a strong activation of the classical (Ang I, Ang II) and alternative RAS (Ang 1-7, Ang 1-5), with highest concentrations found in the HV T group. Although the lung-tissue ACE messenger RNA expression was unchanged, its protein expression showed a dose-dependent increase under mechanical ventilation. The ACE2 messenger RNA expression decreased in all ventilated groups, whereas ACE2 protein levels remained unchanged. Both captopril and Ang 1-7 led to markedly increased Ang 1-7 plasma levels, decreased Ang II levels, and ACE activity (Ang II/Ang I ratio), and effectively prevented VILI. CONCLUSIONS: VILI is accompanied by a strong activation of the RAS. Based on circulating Ang metabolite levels and tissue expression of RAS enzymes, classical ACE-dependent and alternative RAS cascades were activated in the HV T group, whereas classical RAS activation prevailed with VHV T ventilation. Ang 1-7 or captopril protected from VILI primarily by modifying the systemic RAS profile.


Asunto(s)
Sistema Renina-Angiotensina , Lesión Pulmonar Inducida por Ventilación Mecánica , Angiotensina II , Animales , Captopril/metabolismo , Captopril/farmacología , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Sistema Renina-Angiotensina/fisiología , Volumen de Ventilación Pulmonar , Lesión Pulmonar Inducida por Ventilación Mecánica/prevención & control
3.
Anticancer Drugs ; 33(3): 300-307, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34924498

RESUMEN

Small cell lung cancer (SCLC) is frequently found disseminated at first presentation and holds a poor prognosis due to emerging resistance to first-line platinum-based and second-line topotecan chemotherapy. The present investigation tested the antitumor activity of rovalpituzumab tesirine (ROVA-T), a cytotoxic anti-DLL3 drug conjugate, against two SCLC and a corresponding SCLC CTC cell line established from a ROVA-T-resistant patient to characterize the mechanism of recurrence. Two cell lines were established from an SCLC patient progressing under ROVA-T therapy and characterized with respect to chemosensitivity against this drug as well as against currently applied chemotherapeutics and for their delta-like 3 (DLL3) expression. The chemosensitivity assays demonstrate that most SCLC lines show IC50 values exceeding the ROVA-T in-vivo concentrations and that slow-growing cells and lines showing spheroidal growth or proliferation as corresponding circulating tumor cells (CTCs) exhibit higher resistance. Chemosensitivity of the cell lines is not correlated with DLL3 protein expression possibly due to toxicity of the free payload in tissue culture. The clinical trials and experimental results demonstrate that refractoriness to ROVA-T is linked to a low initial tumor expression of DLL3, loss of DLL3 expression, higher chemoresistance to ROVA-T and the putative formation of resistant spheroids by the SCLC cells.


Asunto(s)
Antineoplásicos , Neoplasias Pulmonares , Células Neoplásicas Circulantes , Carcinoma Pulmonar de Células Pequeñas , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Benzodiazepinonas/uso terapéutico , Línea Celular Tumoral , Humanos , Inmunoconjugados , Péptidos y Proteínas de Señalización Intracelular/uso terapéutico , Neoplasias Pulmonares/patología , Proteínas de la Membrana/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología
4.
Int J Mol Sci ; 23(9)2022 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-35562895

RESUMEN

Oxidative phosphorylation is an efficient way to generate the cellular energy currency ATP in a cascade of redox reactions, which ultimately terminate in the reduction of molecular oxygen to water [...].


Asunto(s)
Fosforilación Oxidativa , Oxígeno , Homeostasis , Oxidación-Reducción , Consumo de Oxígeno , Especies Reactivas de Oxígeno
5.
Int J Mol Sci ; 23(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35008532

RESUMEN

Living organisms use a large repertoire of anabolic and catabolic reactions to maintain their physiological body functions, many of which include oxidation and reduction of substrates. The scientific field of redox biology tries to understand how redox homeostasis is regulated and maintained and which mechanisms are derailed in diverse pathological developments of diseases, where oxidative or reductive stress is an issue. The term "oxidative stress" is defined as an imbalance between the generation of oxidants and the local antioxidative defense. Key mediators of oxidative stress are reactive species derived from oxygen, nitrogen, and sulfur that are signal factors at physiological concentrations but can damage cellular macromolecules when they accumulate. However, therapeutical targeting of oxidative stress in disease has proven more difficult than previously expected. Major reasons for this are the very delicate cellular redox systems that differ in the subcellular compartments with regard to their concentrations and depending on the physiological or pathological status of cells and organelles (i.e., circadian rhythm, cell cycle, metabolic need, disease stadium). As reactive species are used as signaling molecules, non-targeted broad-spectrum antioxidants in many cases will fail their therapeutic aim. Precision medicine is called to remedy the situation.


Asunto(s)
Homeostasis/fisiología , Animales , Ritmo Circadiano/fisiología , Humanos , Oxidación-Reducción , Estrés Oxidativo/fisiología , Medicina de Precisión , Transducción de Señal/fisiología
6.
Basic Res Cardiol ; 115(6): 76, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33258993

RESUMEN

Ischemic mitral regurgitation (MR) is a frequent complication of myocardial infarction (MI) characterized by adverse remodeling both at the myocardial and valvular levels. Persistent activation of valvular endothelial cells leads to leaflet fibrosis through endothelial-to-mesenchymal transition (EMT). Tenascin C (TNC), an extracellular matrix glycoprotein involved in cardiovascular remodeling and fibrosis, was also identified in inducing epithelial-to-mesenchymal transition. In this study, we hypothesized that TNC also plays a role in the valvular remodeling observed in ischemic MR by contributing to valvular excess EMT. Moderate ischemic MR was induced by creating a posterior papillary muscle infarct (7 pigs and 7 sheep). Additional animals (7 pigs and 4 sheep) served as controls. Pigs and sheep were sacrificed after 6 weeks and 6 months, respectively. TNC expression was upregulated in the pig and sheep experiments at 6 weeks and 6 months, respectively, and correlated well with leaflet thickness (R = 0.68; p < 0.001 at 6 weeks, R = 0.84; p < 0.001 at 6 months). To confirm the translational potential of our findings, we obtained mitral valves from patients with ischemic cardiomyopathy presenting MR (n = 5). Indeed, TNC was also expressed in the mitral leaflets of these. Furthermore, TNC induced EMT in isolated porcine mitral valve endothelial cells (MVEC). Interestingly, Toll-like receptor 4 (TLR4) inhibition prevented TNC-mediated EMT in MVEC. We identified here for the first time a new contributor to valvular remodeling in ischemic MR, namely TNC, which induced EMT through TLR4. Our findings might set the path for novel therapeutic targets for preventing or limiting ischemic MR.


Asunto(s)
Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Insuficiencia de la Válvula Mitral/metabolismo , Válvula Mitral/metabolismo , Infarto del Miocardio/complicaciones , Tenascina/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Válvula Mitral/patología , Válvula Mitral/fisiopatología , Insuficiencia de la Válvula Mitral/etiología , Insuficiencia de la Válvula Mitral/patología , Insuficiencia de la Válvula Mitral/fisiopatología , Oveja Doméstica , Transducción de Señal , Sus scrofa , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba
7.
Anesth Analg ; 130(2): 321-331, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31498191

RESUMEN

BACKGROUND: Epidural-related maternal fever (ERMF) is an adverse effect of epidural analgesia during labor and is associated with perinatal and neonatal morbidity. Local anesthetics have been proposed to trigger ERMF via sterile inflammation. Ropivacaine is currently the most frequently used epidural anesthetic and considered least toxic. This study investigates molecular effects of ropivacaine on human umbilical vein endothelial cells (HUVECs) as model system for endothelial cells and human placental trophoblasts (TBs), compares the effects to the putative anti-inflammatory lidocaine and investigates the partially alleviating impact of the anti-inflammatory corticosteroid dexamethasone. METHODS: HUVECs and TBs were exposed to ropivacaine (35 µM-7 mM) or lidocaine (21 mM) with or without dexamethasone (1 µM). AnnexinV/propidium iodide staining and lactate dehydrogenase release were used to analyze apoptosis and cytotoxicity. Proinflammatory interleukins-6 (IL-6) and IL-8 as well as prostaglandin E2 (PGE2) were measured by enzyme-linked immunosorbent assay (ELISA), while activation of signaling pathways was detected by Western blotting. Oxidative stress was visualized by live cell imaging and quantification of antioxidant proteins, intercellular adhesion molecule 1, vascular cell adhesion molecule 1, platelet endothelial cell adhesion molecule 1, cyclooxygenase 2, and mitochondrial deoxyribonucleic acid by real-time polymerase chain reaction. Dissipation of the mitochondrial membrane potential was assessed with cytofluorimetric analysis using the J-Aggregate (JC-1 staining [cytofluorimetric analysis using the J-Aggregate]). RESULTS: Ropivacaine exposure dose-dependently induced apoptosis and an increased release of IL-6, IL-8, and PGE2 from HUVECs and TBs. Furthermore, caspase-3, nuclear factor-κB, and p38 mitogen-activated protein kinase pathways were activated, while extracellular signal-regulated kinase 1/2 and protein kinase B (Akt) were dephosphorylated. Downregulation of antioxidative proteins induced oxidative stress and upregulation of ICAM1, VCAM1, and PECAM1 possibly facilitate leukocyte transmigration. Mitochondrial effects included increased release of the proinflammatory mitochondrial DNA damage-associated molecular patterns, but no significant dissipation of the mitochondrial membrane potential. Conversely, lidocaine exhibited repression of IL-6 and IL-8 release over all time points, and early downregulation of COX2 and cell adhesion molecules, which was followed by a late overshooting reaction. Dexamethasone reduced especially inflammatory effects, but as an inducer of mitophagy, had negative long-term effects on mitochondrial function. CONCLUSIONS: This study suggests that ropivacaine causes cellular injury and death in HUVECs and TBs via different signaling pathways. The detrimental effects induced by ropivacaine are only partially blunted by dexamethasone. This observation strengthens the importance of inflammation in ERMF.


Asunto(s)
Anestesia Epidural/efectos adversos , Anestésicos Locales/efectos adversos , Apoptosis/efectos de los fármacos , Fiebre/metabolismo , Mediadores de Inflamación/metabolismo , Ropivacaína/efectos adversos , Anestésicos Locales/administración & dosificación , Apoptosis/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Fiebre/inducido químicamente , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Embarazo , Ropivacaína/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
Eur J Anaesthesiol ; 34(3): 141-149, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28146458

RESUMEN

BACKGROUND: Perioperative oxygen (O2) therapy can cause hyperoxia. Extreme hyperoxia can injure the cardiovascular system and remote organs. OBJECTIVE: Our primary objective was to test the hypothesis that exposure to moderate hyperoxia will induce injury to human umbilical vein endothelial cells (HUVECs), a model for studying the vascular endothelium under controlled conditions. DESIGN: In-vitro cell culture study. SETTING: Department of Anaesthesia, General Intensive Care and Pain Management, Medical University of Vienna, Austria. Study period from the beginning of October 2013 to the end of July 2014. CELLS: HUVECs were isolated from fresh umbilical cords. INTERVENTIONS: HUVECs were exposed to constant hyperoxia (40% O2), cyclic hyperoxia/anoxia (40%/0% O2, average 20% O2), constant normoxia (21% O2) and constant anoxia (0% O2) using a cell culture bioreactor. MAIN OUTCOME MEASURES: Cell growth, viability and release of IL-6, IL-8 and macrophage migration inhibitory factor were assessed at baseline and after 6, 12, 24 and 48 h of treatment. A phosphokinase array was performed after 60 min of treatment to identify activated cellular signalling pathways. RESULTS: Constant hyperoxia and cyclic hyperoxia/anoxia impeded cell growth, reduced viability, triggered a proinflammatory response, proven by IL-6, IL-8 and migration inhibitory factor release, and induced apoptosis and necrosis. The inflammatory and cytotoxicity responses were highest in the constant hyperoxia group. Phosphokinase arrays revealed that different O2 concentrations activated distinct sets of cytoprotective and cell death-associated kinases, including mitogen-activated protein kinases, Src kinases, p53, Akt, mitogen-activated and stress-activated kinase, Lyn, Lck, p70S6, signal transducers and activators of transcription 5b and 6, glycogen synthase kinase 3a/b and 5' AMP-activated protein kinases 1/2. CONCLUSION: Continuous moderate hyperoxia and cyclic moderate hyperoxia/anoxia-induced endothelial inflammation, apoptosis and necrosis. Given the large surface area of the vascular endothelium, moderately elevated O2 levels may contribute to cardiovascular inflammation and injury. TRIAL REGISTRATION: This in-vitro study was not registered in a database.


Asunto(s)
Apoptosis/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Hiperoxia/metabolismo , Mediadores de Inflamación/metabolismo , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Hiperoxia/patología , Inflamación/metabolismo , Inflamación/patología , Necrosis/metabolismo , Necrosis/patología
9.
Anesth Analg ; 122(2): 373-80, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26505576

RESUMEN

BACKGROUND: Intermittent hypoxia may occur in a number of clinical scenarios, including interruption of myocardial blood flow or breathing disorders such as obstructive sleep apnea. Although intermittent hypoxia has been linked to cardiovascular and cerebrovascular disease, the effect of intermittent hypoxia on the human heart is not fully understood. Therefore, in the present study, we compared the cellular responses of cultured human adult cardiac myocytes (HACMs) exposed to intermittent hypoxia and different conditions of continuous hypoxia and normoxia. METHODS: HACMs were exposed to intermittent hypoxia (0%-21% O2), constant mild hypoxia (10% O2), constant severe hypoxia (0% O2), or constant normoxia (21% O2), using a novel cell culture bioreactor with gas-permeable membranes. Cell proliferation, lactate dehydrogenase release, vascular endothelial growth factor release, and cytokine (interleukin [IL] and macrophage migration inhibitory factor) release were assessed at baseline and after 8, 24, and 72 hours of exposure. A signal transduction pathway finder array was performed to determine the changes in gene expression. RESULTS: In comparison with constant normoxia and constant mild hypoxia, intermittent hypoxia induced earlier and greater inflammatory response and extent of cell injury as evidenced by lower cell numbers and higher lactate dehydrogenase, vascular endothelial growth factor, and proinflammatory cytokine (IL-1ß, IL-6, IL-8, and macrophage migration inhibitory factor) release. Constant severe hypoxia showed more detrimental effects on HACMs at later time points. Pathway analysis demonstrated that intermittent hypoxia primarily altered gene expression in oxidative stress, Wnt, Notch, and hypoxia pathways. CONCLUSIONS: Intermittent and constant severe hypoxia, but not constant mild hypoxia or normoxia, induced inflammation and cell injury in HACMs. Cell injury occurred earliest and was greatest after intermittent hypoxia exposure. Our in vitro findings suggest that intermittent hypoxia exposure may produce rapid and substantial damage to the human heart.


Asunto(s)
Hipoxia/complicaciones , Miocarditis/etiología , Miocitos Cardíacos/patología , Adulto , Reactores Biológicos , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Expresión Génica , Humanos , Hipoxia/genética , Hipoxia/patología , L-Lactato Deshidrogenasa/metabolismo , Membranas Artificiales , Miocarditis/genética , Miocarditis/patología , Transducción de Señal , Apnea Obstructiva del Sueño/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(41): 16628-33, 2013 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-24043839

RESUMEN

In schizophrenia, cognitive dysfunction is highly predictive of poor patient outcomes and is not responsive to current medications. Postmortem studies have suggested that cognitive deficits in schizophrenia are correlated with modifications in the number and size of inhibitory synapses. To test if these modifications lead to cognitive deficits, we have created a dominant-negative virus [adeno-associated (AAV)-DN1] that disrupts the clustering of γ-aminobutyric acid type A receptors (GABA(A)Rs) at postsynaptic inhibitory specializations. When injected into the frontal cortex of mice, AAV-DN1 impairs GABA(A)R α2 subunit and GABA transporter 1 (GAT-1) clustering, but increases GABA(A)R α1 subunit clustering on the perisomatic region, with no influence on axon-initial segment clustering. Mice expressing AAV-DN1 have prepulse inhibition deficits and impairments in working memory. Significantly, these behavioral deficits are paralleled by a reduction in electroencephalography γ-power. Collectively, our study provides functional evidence revealing that GABAergic synapses in the prefrontal cortex directly contribute to cognition and γ-power.


Asunto(s)
Cognición/fisiología , Dependovirus/genética , Modelos Animales de Enfermedad , Lóbulo Frontal/metabolismo , Receptores de GABA-A/metabolismo , Esquizofrenia/metabolismo , Animales , Electroencefalografía , Proteínas Transportadoras de GABA en la Membrana Plasmática/metabolismo , Ingeniería Genética/métodos , Vectores Genéticos/genética , Inmunohistoquímica , Ratones , Esquizofrenia/patología , Transducción de Señal/fisiología
11.
Anesth Analg ; 131(2): e87, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-33031681
12.
J Neurosci ; 33(31): 12718-27, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904608

RESUMEN

GABA-mediated tonic and phasic inhibition of thalamic relay neurons of the dorsal lateral geniculate nucleus (dLGN) was studied after ablating tyrosine (Y) phosphorylation of receptor γ2-subunits. As phosphorylation of γ2 Y365 and Y367 reduces receptor internalization, to understand their importance for inhibition we created a knock-in mouse in which these residues are replaced by phenylalanines. On comparing wild-type (WT) and γ2(Y365/367F)+/- (HT) animals (homozygotes are not viable in utero), the expression levels of GABAA receptor α4-subunits were increased in the thalamus of female, but not male mice. Raised δ-subunit expression levels were also observed in female γ2(Y365/367F) +/- thalamus. Electrophysiological analyses revealed no difference in the level of inhibition in male WT and HT dLGN, while both the spontaneous inhibitory postsynaptic activity and the tonic current were significantly augmented in female HT relay cells. The sensitivity of tonic currents to the δ-subunit superagonist THIP, and the blocker Zn(2+), were higher in female HT relay cells. This is consistent with upregulation of extrasynaptic GABAA receptors containing α4- and δ-subunits to enhance tonic inhibition. In contrast, the sensitivity of GABAA receptors mediating inhibition in the female γ2(Y356/367F) +/- to neurosteroids was markedly reduced compared with WT. We conclude that disrupting tyrosine phosphorylation of the γ2-subunit activates a sex-specific increase in tonic inhibition, and this most likely reflects a genomic-based compensation mechanism for the reduced neurosteroid sensitivity of inhibition measured in female HT relay neurons.


Asunto(s)
Cuerpos Geniculados/citología , Inhibición Neural/fisiología , Neuronas/fisiología , Receptores de GABA-A/metabolismo , Tirosina/metabolismo , Anestésicos/farmacología , Animales , Animales Recién Nacidos , Línea Celular Transformada , Femenino , Cuerpos Geniculados/fisiología , Humanos , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Fosforilación , Receptores de GABA-A/genética , Receptores de GABA-B/química , Receptores de GABA-B/genética , Receptores de GABA-B/metabolismo , Tirosina/genética
13.
J Neurosci ; 33(17): 7264-73, 2013 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-23616535

RESUMEN

Intravenous anesthetics exert a component of their actions via potentiating inhibitory neurotransmission mediated by γ-aminobutyric type-A receptors (GABAARs). Phasic and tonic inhibition is mediated by distinct populations of GABAARs, with the majority of phasic inhibition by subtypes composed of α1-3ßγ2 subunits, whereas tonic inhibition is dependent on subtypes assembled from α4-6ßδ subunits. To explore the contribution that these distinct forms of inhibition play in mediating intravenous anesthesia, we have used mice in which tyrosine residues 365/7 within the γ2 subunit are mutated to phenyalanines (Y365/7F). Here we demonstrate that this mutation leads to increased accumulation of the α4 subunit containing GABAARs in the thalamus and dentate gyrus of female Y365/7F but not male Y365/7F mice. Y365/7F mice exhibited a gender-specific enhancement of tonic inhibition in the dentate gyrus that was more sensitive to modulation by the anesthetic etomidate, together with a deficit in long-term potentiation. Consistent with this, female Y365/7F, but not male Y365/7F, mice exhibited a dramatic increase in the duration of etomidate- and propofol-mediated hypnosis. Moreover, the amnestic actions of etomidate were selectively potentiated in female Y365/7F mice. Collectively, these observations suggest that potentiation of tonic inhibition mediated by α4 subunit containing GABAARs contributes to the hypnotic and amnestic actions of the intravenous anesthetics, etomidate and propofol.


Asunto(s)
Amnesia/inducido químicamente , Etomidato/administración & dosificación , Hipnóticos y Sedantes/administración & dosificación , Potenciación a Largo Plazo/efectos de los fármacos , Inhibición Neural/efectos de los fármacos , Propofol/administración & dosificación , Amnesia/fisiopatología , Anestésicos Intravenosos/administración & dosificación , Animales , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/fisiología , Técnicas de Cultivo de Órganos , Distribución Aleatoria
14.
Front Physiol ; 14: 1109452, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064885

RESUMEN

Introduction: Ventilator-induced lung injury (VILI) may aggravate critical illness. Although angiotensin-converting enzyme (ACE) inhibition has beneficial effects in ventilator-induced lung injury, its clinical application is impeded by concomitant hypotension. We hypothesized that the aminopeptidase inhibitor ALT-00 may oppose the hypotension induced by an angiotensin-converting enzyme inhibitor, and that this combination would activate the alternative renin-angiotensin system (RAS) axis to counteract ventilator-induced lung injury. Methods: In separate experiments, C57BL/6 mice were mechanically ventilated with low (LVT, 6 mL/kg) and high tidal volumes (HVT, 30 mL/kg) for 4 h or remained unventilated (sham). High tidal volume-ventilated mice were treated with lisinopril (0.15 µg/kg/min) ± ALT-00 at 2.7, 10 or 100 µg/kg/min. Blood pressure was recorded at baseline and after 4 h. Lung histology was evaluated for ventilator-induced lung injury and the angiotensin (Ang) metabolite profile in plasma (equilibrium levels of Ang I, Ang II, Ang III, Ang IV, Ang 1-7, and Ang 1-5) was measured with liquid chromatography tandem mass spectrometry at the end of the experiment. Angiotensin concentration-based markers for renin, angiotensin-converting enzyme and alternative renin-angiotensin system activities were calculated. Results: High tidal volume-ventilated mice treated with lisinopril showed a significant drop in the mean arterial pressure at 4 h compared to baseline, which was prevented by adding ALT-00 at 10 and 100 µg/kg/min. Ang I, Ang II and Ang 1-7 plasma equilibrium levels were elevated in the high tidal volumes group versus the sham group. Lisinopril reduced Ang II and slightly increased Ang I and Ang 1-7 levels versus the untreated high tidal volumes group. Adding ALT-00 at 10 and 100 µg/kg/min increased Ang I and Ang 1-7 levels versus the high tidal volume group, and partly prevented the downregulation of Ang II levels caused by lisinopril. The histological lung injury score was higher in the high tidal volume group versus the sham and low tidal volume groups, and was attenuated by lisinopril ± ALT-00 at all dose levels. Conclusion: Combined angiotensin-converting enzyme plus aminopeptidase inhibition prevented systemic hypotension and maintained the protective effect of lisinopril. In this study, a combination of lisinopril and ALT-00 at 10 µg/kg/min appeared to be the optimal approach, which may represent a promising strategy to counteract ventilator-induced lung injury that merits further exploration.

15.
J Neurosci ; 31(41): 14677-87, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21994384

RESUMEN

The majority of fast synaptic inhibition in the brain is mediated by benzodiazepine-sensitive α1-subunit-containing GABA type A receptors (GABA(A)Rs); however, our knowledge of the mechanisms neurons use to regulate their synaptic accumulation is rudimentary. Using immunoprecipitation, we demonstrate that GABA(A)Rs and gephyrin are intimately associated at inhibitory synapses in cultured rat neurons. In vitro we reveal that the E-domain of gephyrin directly binds to the α1 subunit with an affinity of ∼20 µm, mediated by residues 360-375 within the intracellular domain of this receptor subunit. Mutating residues 360-375 decreases both the accumulation of α1-containing GABA(A)Rs at gephyrin-positive inhibitory synapses in hippocampal neurons and the amplitude of mIPSCs. We also demonstrate that the affinity of gephyrin for the α1 subunit is modulated by Thr375, a putative phosphorylation site. Mutation of Thr375 to a phosphomimetic, negatively charged amino acid decreases both the affinity of the α1 subunit for gephyrin, and therefore receptor accumulation at synapses, and the amplitude of mIPSCs. Finally, single-particle tracking reveals that gephyrin reduces the diffusion of α1-subunit-containing GABA(A)Rs specifically at inhibitory synapses, thereby increasing their confinement at these structures. Our results suggest that the direct binding of gephyrin to residues 360-375 of the α1 subunit and its modulation are likely to be important determinants for the stabilization of GABA(A)Rs at synaptic sites, thereby modulating the strength of synaptic inhibition.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Inhibición Neural/fisiología , Receptores de GABA-A/metabolismo , Sinapsis/metabolismo , Animales , Calorimetría/métodos , Proteínas Portadoras/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos , Femenino , Hipocampo/citología , Potenciales Postsinápticos Inhibidores/genética , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Proteínas de la Membrana/genética , Ratones , Microscopía Confocal , Mutación , Neuronas/clasificación , Neuronas/fisiología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Técnicas de Placa-Clamp , Unión Proteica/genética , Unión Proteica/fisiología , Ratas , Receptores de GABA-A/genética , Treonina/genética , Treonina/metabolismo , Transfección/métodos , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas
16.
J Biol Chem ; 286(49): 42105-42114, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22006921

RESUMEN

Gephyrin is the major protein determinant for the clustering of inhibitory neurotransmitter receptors. Earlier analyses revealed that gephyrin tightly binds to residues 398-410 of the glycine receptor ß subunit (GlyR ß) and, as demonstrated only recently, also interacts with GABA(A) receptors (GABA(A)Rs) containing the α1, α2, and α3 subunits. Here, we dissect the molecular basis underlying the interactions between gephyrin and GABA(A)Rs containing these α-subunits and compare them to the crystal structure of the gephyrin-GlyR ß complex. Biophysical and biochemical assays revealed that, in contrast to its tight interaction with GlyR ß, gephyrin only loosely interacts with GABA(A)R α2, whereas it has an intermediate affinity for the GABA(A)R α1 and α3 subunits. Despite the wide variation in affinities and the low overall sequence homology among the identified receptor subunits, competition assays confirmed the receptor-gephyrin interaction to be a mutually exclusive process. Selected gephyrin point mutants that critically weaken complex formation with GlyR ß also abolished the GABA(A)R α1 and α3 interactions. Additionally, we identified a common binding motif with two conserved aromatic residues that are central for gephyrin binding. Consistent with the biochemical data, mutations of the corresponding residues within the cytoplasmic domain of α2 subunit-containing GABA(A)Rs attenuated clustering of these receptors at postsynaptic sites in hippocampal neurons. Taken together, our experiments provide key insights regarding similarities and differences in the complex formation between gephyrin and GABA(A)Rs compared with GlyRs and, hence, the accumulation of these receptors at postsynaptic sites.


Asunto(s)
Proteínas Portadoras/química , Proteínas de la Membrana/química , Receptores de GABA-A/metabolismo , Receptores de Glicina/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Cloruros/química , Análisis por Conglomerados , Ligandos , Modelos Moleculares , Conformación Molecular , Mutación , Unión Proteica , Mapeo de Interacción de Proteínas , Ratas , Sinapsis/metabolismo , Potenciales Sinápticos , Tirosina/química
17.
J Biol Chem ; 286(43): 37702-11, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21880742

RESUMEN

The multifunctional scaffolding protein gephyrin is a key player in the formation of the postsynaptic scaffold at inhibitory synapses, clustering both inhibitory glycine receptors (GlyRs) and selected GABA(A) receptor (GABA(A)R) subtypes. We report a direct interaction between the GABA(A)R α3 subunit and gephyrin, mapping reciprocal binding sites using mutagenesis, overlay, and yeast two-hybrid assays. This analysis reveals that critical determinants of this interaction are located in the motif FNIVGTTYPI in the GABA(A)R α3 M3-M4 domain and the motif SMDKAFITVL at the N terminus of the gephyrin E domain. GABA(A)R α3 gephyrin binding-site mutants were unable to co-localize with endogenous gephyrin in transfected hippocampal neurons, despite being able to traffic to the cell membrane and form functional benzodiazepine-responsive GABA(A)Rs in recombinant systems. Interestingly, motifs responsible for interactions with GABA(A)R α2, GABA(A)R α3, and collybistin on gephyrin overlap. Curiously, two key residues (Asp-327 and Phe-330) in the GABA(A)R α2 and α3 binding sites on gephyrin also contribute to GlyR ß subunit-E domain interactions. However, isothermal titration calorimetry reveals a 27-fold difference in the interaction strength between GABA(A)R α3 and GlyR ß subunits with gephyrin with dissociation constants of 5.3 µm and 0.2 µm, respectively. Taken together, these observations suggest that clustering of GABA(A)R α2, α3, and GlyRs by gephyrin is mediated by distinct mechanisms at mixed glycinergic/GABAergic synapses.


Asunto(s)
Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de GABA-A/metabolismo , Sinapsis/metabolismo , Secuencias de Aminoácidos , Animales , Proteínas Portadoras/genética , Membrana Celular/genética , Neuronas GABAérgicas/citología , Células HEK293 , Hipocampo/citología , Humanos , Proteínas de la Membrana/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Ratas , Receptores de GABA-A/genética , Sinapsis/genética
18.
Proc Natl Acad Sci U S A ; 106(47): 20039-44, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19903874

RESUMEN

Fast synaptic inhibition in the brain is largely mediated by gamma-aminobutyric acid receptors (GABA(A)R). While the pharmacological manipulation of GABA(A)R function by therapeutic agents, such as benzodiazepines can have profound effects on neuronal excitation and behavior, the endogenous mechanisms neurons use to regulate the efficacy of synaptic inhibition and their impact on behavior remains poorly understood. To address this issue, we created a knock-in mouse in which tyrosine phosphorylation of the GABA(A)Rs gamma2 subunit, a posttranslational modification that is critical for their functional modulation, has been ablated. These animals exhibited enhanced GABA(A)R accumulation at postsynaptic inhibitory synaptic specializations on pyramidal neurons within the CA3 subdomain of the hippocampus, primarily due to aberrant trafficking within the endocytic pathway. This enhanced inhibition correlated with a specific deficit in spatial object recognition, a behavioral paradigm dependent upon CA3. Thus, phospho-dependent regulation of GABA(A)R function involving just two tyrosine residues in the gamma2 subunit provides an input-specific mechanism that not only regulates the efficacy of synaptic inhibition, but has behavioral consequences.


Asunto(s)
Hipocampo/metabolismo , Memoria/fisiología , Receptores de GABA-A/metabolismo , Conducta Espacial/fisiología , Tirosina/metabolismo , Animales , Técnicas de Sustitución del Gen , Hipocampo/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Técnicas de Placa-Clamp , Fosforilación , Receptores de GABA-A/genética
19.
Antioxidants (Basel) ; 11(12)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36552557

RESUMEN

Patients presenting with insufficient tissue oxygenation and impaired lung function as in acute respiratory distress syndrome (ARDS) frequently require mechanical ventilation with supplemental oxygen. Despite the lung being used to experiencing the highest partial pressure of oxygen during healthy breathing, the organ is susceptible to oxygen-induced injury at supraphysiological concentrations. Hyperoxia-induced lung injury (HALI) has been regarded as a second hit to pre-existing lung injury and ventilator-induced lung injury (VILI) attributed to oxidative stress. The injured lung has a tendency to form atelectasis, a cyclic collapse and reopening of alveoli. The affected lung areas experience oxygen conditions that oscillate between hyperoxia and hypoxia rather than remaining in a constant hyperoxic state. Mechanisms of HALI have been investigated in many animal models previously. These studies provided insights into the effects of hyperoxia on the whole organism. However, cell type-specific responses have not been dissected in detail, but are necessary for a complete mechanistic understanding of ongoing pathological processes. In our study, we investigated the effects of constant and intermittent hyperoxia on the lung endothelium from a mouse by an in vitro proteomic approach. We demonstrate that these oxygen conditions have characteristic effects on the pulmonary endothelial proteome that underlie the physiological (patho)mechanisms.

20.
Mol Cell Neurosci ; 44(2): 129-34, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20233604

RESUMEN

Phosphorylation of GABA(A) receptors is an important mechanism for dynamically modulating inhibitory synaptic function in the mammalian brain. In particular, phosphorylation of tyrosine residues 365 and 367 (Y365/7) within the GABA(A) receptor gamma2 subunit negatively regulates the endocytosis of GABA(A) receptors and enhances synaptic inhibition. Here we show that Fyn, a Src family kinase (SFK), interacts with the gamma2 subunit in a phosphorylation-dependent manner. Furthermore, we demonstrate that Fyn binds within a region of the gamma2 intracellular domain that is centered on residues Y365/7, with the phosphorylation of Y367 being particularly important for mediating this interaction. Tyrosine phosphorylation of the gamma2 subunit is significantly reduced in the hippocampus of Fyn knockout mice, suggesting that Fyn is an important kinase that contributes to the phosphorylation of this subunit in vivo. Tyrosine phosphorylation of the gamma2 subunit is not completely abolished in Fyn kinase mice, suggesting that other SFKs, such as Src, also contribute to maintaining and regulating the endogenous phosphorylation level of gamma2-containing GABA(A) receptors. In summary, we demonstrate Fyn as one of the SFKs that binds to and phosphorylates the gamma2 subunit of the GABA(A) receptor. This has important implications for the regulation of synaptic GABA(A) receptors via signaling pathways that lead to the activation of Fyn kinase.


Asunto(s)
Encéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Receptores de GABA-A/metabolismo , Secuencia de Aminoácidos/fisiología , Animales , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fosforilación , Estructura Terciaria de Proteína/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/química , Transducción de Señal/fisiología , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA